You are on page 1of 16

Acta Neuropathologica (2018) 136:41–56

https://doi.org/10.1007/s00401-018-1868-1

ORIGINAL PAPER

Alzheimer’s disease pathology propagation by exosomes containing


toxic amyloid‑beta oligomers
Maitrayee Sardar Sinha1 · Anna Ansell‑Schultz1 · Livia Civitelli1 · Camilla Hildesjö1 · Max Larsson1 · Lars Lannfelt2,3 ·
Martin Ingelsson2 · Martin Hallbeck1 

Received: 20 December 2017 / Revised: 15 May 2018 / Accepted: 19 May 2018 / Published online: 13 June 2018
© The Author(s) 2018

Abstract
The gradual deterioration of cognitive functions in Alzheimer’s disease is paralleled by a hierarchical progression of amy-
loid-beta and tau brain pathology. Recent findings indicate that toxic oligomers of amyloid-beta may cause propagation of
pathology in a prion-like manner, although the underlying mechanisms are incompletely understood. Here we show that
small extracellular vesicles, exosomes, from Alzheimer patients’ brains contain increased levels of amyloid-beta oligomers
and can act as vehicles for the neuron-to-neuron transfer of such toxic species in recipient neurons in culture. Moreover,
blocking the formation, secretion or uptake of exosomes was found to reduce both the spread of oligomers and the related
toxicity. Taken together, our results imply that exosomes are centrally involved in Alzheimer’s disease and that they could
serve as targets for development of new diagnostic and therapeutic principles.

Keywords  Alzheimer’s disease · Exosomes · Oligomers · Beta-amyloid · Human · Prion-like · Propagation

Introduction 38–40, 61] as well as spread of tau between neurons in the


brain of transgenic mouse models [25]. These observations
Gradual accumulation of toxic amyloid-beta (Aβ) and tau are are further supported by the distribution of Aß and tau PET
believed to be central to Alzheimer´s disease (AD) patho- ligands in relation to the brain connectome [45].
genesis. These abnormal deposits typically appear with a Different Aβ conformational states have different proper-
hierarchical spatial distribution, which suggest that the ties, and intermediate products of fibril formation, such as
pathological proteins can propagate between different brain lower molecular weight Aβ oligomers (oAβ) and protofi-
areas [7, 52, 53]. Accordingly, recent studies have demon- brils, have been suggested to be particularly neurotoxic and
strated transfer of Aβ in cellular and animal models [27, to act as seeds for further aggregation [27, 61]. In addition,
these soluble forms of Aß correlate better than fibrils with
cognitive function in the AD brain [39]. We have previ-
Maitrayee Sardar Sinha and Anna Ansell-Schultz are equal ously shown that oAβ can accumulate inside cells and sub-
contributors.
sequently spread from one cell to another [15, 40]. These
Electronic supplementary material  The online version of this findings thus suggest that Aβ can propagate pathology in
article (https​://doi.org/10.1007/s0040​1-018-1868-1) contains a manner similar to what is seen for prion disorders and
supplementary material, which is available to authorized users. for several other neurodegenerative diseases [8]. However,
the underlying mechanisms for the spreading of toxic oAβ
* Martin Hallbeck
Martin.Hallbeck@liu.se remain incompletely understood [28].
Exosomes, small extracellular vesicles (20–120 nm in diam-
1
Department of Pathology, Department of Clinical eter) [58] developing from endosomes through multivesicular
and Experimental Medicine, Linköping University, bodies, have recently emerged as key players in cellular com-
Linköping, Sweden
munication and transport of molecules in both health and dis-
2
Department of Public Health and Caring Sciences, ease, including neuronal toxicity [33] and neurodegenerative
Geriatrics, Uppsala University, Uppsala, Sweden
disorders [59]. Exosomes can carry different cargos such as
3
BioArctic AB, Warfvinges väg 25, 112 85 Stockholm, proteins, RNA and miRNA and can also contain monomeric
Sweden

13
Vol.:(0123456789)

42 Acta Neuropathologica (2018) 136:41–56

Aß, tau and α-synuclein [12, 17, 46, 59] and can propagate tau described [6]. After 5 min blocking of endogenous peroxi-
pathology [4]. Thus, exosomes could potentially also carry dase by incubation in Background Sniper (Biocare Medical),
aggregated proteins such as oAß. However, no study has so far slides were washed in Tris-buffered saline (TBS) solution
investigated if exosomes isolated from AD brain tissue can be and incubated with rabbit polyclonal anti-flotillin-1 (Abcam,
responsible for interneuronal protein transfer. 1:200) followed by incubation with either the second pri-
For the first time, we now demonstrate that intracellular mary mouse monoclonal antibody (mAb158, 1:7500, Bio-
oAβ is co-localized with exosomes and show that AD brain- Arctic) or mouse monoclonal antibody 82E1 (1:25, IBL
derived exosomes can mediate neuron-to-neuron propaga- International) for 30 min at room temperature (RT).
tion of oAß. Furthermore, we show that the concentration of After rinsing in TBS, the slides were incubated for 30 min
oAβ in exosomes is significantly increased in post mortem with MACH 2 Double Stain (Biocare Medical). Following
AD brains. In addition, exosomes carrying oAβ can be inter- this step, a 30 min incubation was performed with an AP
nalized in cultured neurons and spread their toxic content to linked chromogen IP Warp red/HRP linked chromogen Vina
nearby cells. Green cocktail (Biocare Medical). After rinsing in deionized
water, the sections were counterstained with Mayers haema-
toxylin (Histolab Products AB) and mounted with Pertex
Methods mounting media (Histolab Products AB) and micrographs
were obtained using 100 × oil immersion objective (Nikon
Brain tissues Eclipse 80i, Digital Sight DS-Fi1).
After blocking and incubating with primary antibod-
Post  mortem brain samples of temporal neocortex from ies as described above, the slides were rinsed in TBS and
healthy control (1 female, 4 male) and AD (4 female, 1 male) incubated with a Fluorescence Enhancement Probe Mouse/
subjects (Table 1) were provided by the brain bank at Uppsala Fluorescence Enhancement Probe Rabbit cocktail (Biocare
University, Sweden. The tissue was received as fresh-frozen Medical) for 20 min subsequent to a 40 min incubation with
or as formalin‐fixed (4% formaldehyde) paraffin-embedded a Goat-anti-Mouse DyLight 549/Goat Anti-Rabbit DyLight
blocks. The AD cases were neuropathologically diagnosed 488 cocktail (Biocare Medical) diluted 1:200, respectively.
as CERAD C, Braak stages IV–VI. None of the control After rinsing in deionized water, the sections were nuclear
patients suffered from dementia or any neurodegenerative counterstained with DAPI and mounted with Fluoro Care
disorder. The collection and use of post mortem brain tissue Anti-Fade Mountant (Biocare Medical) and analysed with a
was approved by the Regional Ethical Committee in Uppsala, Zeiss LSM 700 confocal microscope. The differential inter-
Sweden (2005/103, 2005-06-29; 2009/089; 2009-04-22). face contrast (DIC) mode and 405, 488, 555 and 639 lasers
were used to acquire the images with 63x/1.40 oil immersion
Immunohistochemistry and immunofluorescence plan-apochromatic DICII objectives. The micrographs were
of brain sections processed using Huygens (Scientific Volume Imaging) and
ZEN lite (blue edition) software.
Formalin-fixed, paraffin-embedded 10 μm sections of tempo-
ral neocortex from healthy control and AD brains (Table 1)
were used for this study and pretreated as previously

Table 1  Demographic and Diagnosis Age Sex PMI Braak Thal CERAD NIA-Reagan αSyn
clinical characteristics of
post mortem cases used in the AD 61 F 48 5–6 5 C High prob None
study
AD 64 M 12 5–6 5 C High prob None
AD 85 F 21 5–6 3 B High prob None
AD 90 F 11 5 3 B None
AD 63 F 48 6 5 C None
C 88 M 39 0 0 0 None
C 88 F 22 2 0 0 In S Nigra
C 63 M 30 0 0 0 None
C 90 M 30 0 1 0 None
C 91 M 27 3 4 C None

5 severe Alzheimer’s disease cases  (Braak stages V–VI) and 5 nondemented cases  (Braak stages 0–III)
were used to determine the presence of oAβ in brain derived exosomes

13
Acta Neuropathologica (2018) 136:41–56 43

Cell lines and differentiation in a molar ratio of 2:, incubated overnight at 4 °C. Labelled
Aβ1-42 was diluted to a final concentration of 100 µM in
Two different cultured cell types were used in the study: the HEPES 20 mM pH 7.4, vortexed, sonicated for 10 min and
human-induced pluripotent stem cells, AF22 (from a control incubated at 4 °C overnight. After the overnight incubation,
subject) and the human neuroblastoma cell line, SH-SY5Y Aβ1-42 was separated from free dye with size exclusion
(ECACC: Sigma-Aldrich). The neuroepithelial stem cell chromatography (SEC). A Sephadex 75 10/300 GL column
line, AF22, derived from human-induced pluripotent stem coupled to a liquid chromatography system (ÄKTA pure,
cells from human skin fibroblasts was provided by Dr. Anna GE Healthcare) was equilibrated with ­NH4HCO3 50 mM
Falk, Karolinska Institute, Sweden. The process of repro- pH 8.5 and 500 µl of sample was injected into the column.
gramming human cells was approved by the Ethical Com- To estimate the molecular weight of the Aβ species, LMW
mittee at Karolinska Institute, Sweden (dnr 2012/208-31/3 gel filtration calibration kits (GE Healthcare) were used.
with addendum 2012/856-32). All samples were given with Oligomeric and monomeric Aβ species were eluted at a
informed consent. The AF22 cell line has previously been flow rate of 0.5 ml/min, collected and lyophilized. Then,
shown to have a stable neuronal differentiation competence Aβ species (oAβ-AF700) were resuspended in phosphate-
and the capacity to generate functionally mature human buffered saline (PBS) solution and quantified spectrophoto-
neurons [20], denoted hiPSC. The hiPSCs were cultured metrically at 215 nm by using the Aβ1-42 extinction coef-
on 0.01% poly-l-ornithine and laminin (10 µg/mL, Sigma- ficient (Aβ1-42 ɛ214 nm = 75,887 M−1 cm−1) according to
Aldrich) coated cell culture flasks (Corning) in DMEM/ Lambert–Beer’s law. Protein aliquots were stored at − 80 °C.
F12 media (Gibco by Life Technologies), supplemented
with EGF (10 ng/mL, PeproTech), FGF2 (10 ng/mL, Pep- Exosome purification, characterization and labelling
roTech), N2 (5 µl/ml, Life Technologies) and B27 (1 ml/L,
Life Technologies) and further differentiated for 40 days to Isolation of brain exosomes from extracellular space of
functionally mature human neurons in 1:1 DMEM, neuroba- freshly frozen human brain tissues (250 mg) was performed
sal media containing B27 (10 µl/ml), Laminin (1 µl/ml) and as previously described [42]. Tissue was dissociated with
N2 (5 µl/ml) as previously described [20]. papain (20 units/ml, 15 min at 37 °C, Sigma-Aldrich) fol-
Neuronal differentiation of the human neuroblastoma cell lowed by filtration through 40 µm mesh filter (BD Bio-
line SH-SY5Y was performed as previously described [1]. sciences) and a 0.2 µm syringe filter (Thermo Scientific) to
In brief, SH-SY5Y cells were cultured and pre-differentiated separate extracellular matrix from cells. The crude exosomes
for 7 days using 10 μM retinoic acid (RA; Sigma-Aldrich; were then isolated by differential centrifugation method and
denoted as raSH-SY5Y). Pre-differentiated raSH-SY5Y subsequently purified by sucrose density gradient as previ-
cells were seeded on 6-, 12- or 24-well glass plates coated ously described and resuspended in PBS, lysis buffer or dilu-
with 20% extracellular matrix (ECM) gel (BD Bioscience) ent C (Sigma-Aldrich) for further experiments [42].
and further differentiated for 10 days with serum-free MEM Exosomes from conditioned media of raSH-SY5Y cells
(Gibco by Life Technologies) supplemented with brain- were isolated by differential ultracentrifugation. In brief,
derived neurotrophic factor (BDNF, 50 ng/ml, PeproTech), 50–80 million raSH-SY5Y cells were incubated with oAβ-
neuregulin β1 (NRGβ1,10 ng/ml, R&D Systems), nerve AF700for 3 h at 37 °C. After PBS washing, cells were kept
growth factor (NGF, 10 ng/ml, R&D Systems) and vitamin for 48 h in MEM supplemented with exosome-free serum
D3 (VitD3, 24 nM, Sigma-Aldrich). These fully differenti- (System Biosciences). Culture supernatants were collected
ated cells are denoted dSH-SY5Y. and spun at 1000 × g for 10 min for removal of cellular
In addition, SH-SY5Y cells expressing a CD63-EGFP debris. The supernatants were then filtered through 0.22 µM
fusion protein were generated using AddGene plasmid filter and sequentially centrifuged at 5000×g, 10,000×g, and
#62964. 100,000×g. The final pellet was then resuspended in PBS,
lysis buffer or diluent C for further analysis.
Labelling and oligomerization of Aβ1‑42 Exosomes were labelled with PKH67 or PKH26 dye
(Sigma-Aldrich), according to the manufacturer’s protocol.
Recombinant Aβ1-42 peptides (Innovagen,) was dissolved Briefly, 4 μL PKH67 dye was mixed with exosome suspen-
in 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP, Sigma-Aldrich) sion in diluent C and incubated for 10 min at 37 °C. The
and vacuum dried overnight. Aβ1-42 (1.054 mM final con- labelling reaction was stopped by adding 20 ml chilled PBS.
centration) was resuspended in N ­ a2CO3 (0.1 M pH 8.5) and Labelled exosomes were ultra-centrifuged at 100,000×g
incubated with the fluorophore Alexa Fluor 700 (AF700) for 70 min, washed with PBS, ultra-centrifuged again at
succinimidyl ester (1.58 mM final concentration, Life Tech- 100,000×g and the pellet was resuspended in PBS.
nologies) for 40 min at 4 °C or the fluorophore 6-carboxyte-
tramethylrhodamine succinimidyl ester (TMR, Invitrogen),

13

44 Acta Neuropathologica (2018) 136:41–56

Cellular uptake of exosomes flow cytometry or fixed with 4% PFA for immunofluorescent
staining. Additionally, to control for donor cell contamina-
dSH-SY5Y or hiPSCs cells were plated on coverslips in the tion in the recipient cell samples, donor cells were trans-
respective serum-free growth medium. Before the uptake fected with BacMam 2.0 early endosomes Rab5a-RFP (Life
assay, exosomes were isolated from brain tissue or condi- Technologies) at a final concentration of 30 particles per cell
tioned media of raSH-SY5Y cells and labelled with PKH67 before co-culture and RFP fluorescence was monitored in
or PKH26 as described above. In order to use equal amounts recipient cells by flow cytometry.
of exosomes in the cell cultures, exosomal protein content
was quantitated by using BCA (Bio-Rad) or QuantIT (Inv- Immunocytochemistry
itrogen). Brain exosome abundance was quantified accord-
ing to the AChE activity (EXOCET Exosome Quantification Co-localization of oAβ and flotillin-1 and TSG101 were
kit; System Biosciences) according to the manufacturer’s visualized with immunostaining using 1: 5000 solution of
protocol. The uptake was performed by incubating cell cul- mouse anti-mAb158, 1: 200 solution of mouse anti-flotil-
tures with 100 µl of exosome solutions (corresponding to an lin-1 and a 1: 200 solution of mouse anti-TSG101. The sec-
exosomal protein content of 0.62 ± 0.28 μg/μl from brain or ondary antibodies were Cy3 conjugated (Jackson Immuno
0.71 ± 0.33 μg/μl from conditioned media; equal to 1.4e10 Research, 1:1000) and Alexa Fluor 488-conjugated (Invit-
exosome abundance from brain) in a humid chamber for 3 h rogen, 1:400) goat anti-mouse IgG. GFP was detected using
(37 °C, 5% C­ O2). For inhibition experiments, cultured cells 1:200 rabbit anti-GFP (Life Technologies) and 1:400 goat
were pre-incubated for 30 min with the endocytosis inhibi- anti rabbit Alexa flour 647 (Life Technologies).
tors, dynasore (dynamin inhibitor, 80 µM), phenylarsine
oxide (clathrin inhibitor 20 µM), genistein (caveolae inhibi- Cell microscopy
tor, 200 µM) all from Sigma-Aldrich. Isolated exosomes in
PBS were added to cells for 3 h as above and flow cytometry Images of fixed cells were acquired with a Zeiss LSM 700
was performed. confocal microscope. The differential interface contrast
(DIC) mode and 405, 488, 555 and 639 lasers were used
Co‑culture model to acquire the images with 63x/1.40 oil immersion plan-
apochromatic DICII objectives. Live cell imaging were done
Co-culture of donor-recipient cells was performed by using using a Zeiss Primo Vert microscope. The micrographs were
two different methods namely the coverslip system (where processed using Huygens (Scientific Volume Imaging) and
physical contact of synapses is possible) or the transwell ZEN lite (blue edition) software.
system (where physical contact of synapses is not possible).
In both cases, donor cells (raSH-SY5Y 12,500 cells/cm2, Flow cytometry
or hiPSCs 25,000 cells/cm2) were seeded on glass cover-
slips coated with 0.1 mg/ml poly-l-ornithine and 10 µg/ml To detect PKH67 labelled exosomes or oAβ-AF700, cells
laminin and cultured as described above for 3 h at 37 °C were released from the ECM gel using Corning Recovery
with either 1 µM of oAβ-AF700 or labelled exosomes from Solution (Corning) according to manufacturer’s instruc-
brain tissue or conditioned media, and thereafter washed tions, filtered through CellTrics 30 µm filters (Sysmex),
twice with PBS. re-suspended in PBS, and subsequently analysed on a BD
In the transwell system, donor cells were seeded on a FACSAria™ (BD Biosciences) flow cytometer.
polycarbonate membrane filter with a 0.4 µm pore size (Fal- After inhibiting the gene expression of exosome mark-
con, Corning), placed on top of recipient cells (dSH-SY5Y) ers TSG101 and VPS4A in raSH-SY5Y cells, using RNA
and subsequently co-cultured for 24 h. At the end of incuba- interference, the number of secreted exosomes were ana-
tion, the membrane filter was removed and recipient cells lysed using the Exo-Flow™ kit (System Biosciences, USA),
were washed with PBS and analysed with flow cytometry or targeting CD9, CD63 and CD81, as per manufacture’s
fixed with 4% PFA for immunofluorescent labelling. instruction.
In the coverslip system, the donor cells were seeded on
glass coverslips (VWR International) and placed upside Enzyme‑linked immunosorbent assay (ELISA)
down on top of recipient cells, predifferentiated as described analysis
above (resulting in donor cells facing recipient cells) and
subsequently co-cultured for 24 or 48 h at 37 °C. For gel Altogether, 96-well EIA/RIA plates (Corning Inc.) were
cultured cells this results in a 3D environment. Thereafter, coated at 4 °C overnight with 200 ng/well of mAb158 in
the coverslips with donor cells were removed and recipi- PBS. Plates were blocked with 1% bovine serum albumin
ent cells were washed with PBS and either analysed with (BSA) in TBS. Exosome samples prepared in RIPA buffer

13
Acta Neuropathologica (2018) 136:41–56 45

(150 mM sodium chloride, 1% Triton X-100, 0.5% sodium (Thermo Scientific) detection systems and analysed by
deoxycholate, 0.1% sodium dodecyl sulfate and 50 mM Tris, ImageJ software.
pH 8.0) were added to the plates in duplicates and incu-
bated for 2 h at 37 °C. A total of 1 mg/mL of biotinylated Negative staining and transmission electron
mAb158 was added and incubated for 1 h at 22 °C, followed microscopy of exosomes
by 1 h at RT incubation of streptavidin-coupled poly-HRP
(Mabtech). K-blue enhanced (ANL product, Sweden) was Exosome suspensions were fixed in 4% paraformaldehyde
used as HRP substrate and plates were read in a spectropho- (at 1:1 dilution, for a final paraformaldehyde concentration
tometer at 450 nm, using Spectra MAX 190 and then ana- of 2%) overnight at 4 °C and stored at − 20 °C until use.
lysed with SOFT Max Pro. Wells were washed three times Thawed exosome suspensions were vortexed briefly and
in TBST between each step. The 82E1 sandwich ELISA centrifuged in a microcentrifuge for 30 s. Exosomes were
was performed same as above using 0.25 µg/ml for capture adsorbed on Formvar-coated Ni mesh grids by placing the
and detection antibody [56]. The amount of oAβ within grids on 5 µl drops of exosome suspension for 20 min in a
exosomes was quantified with respect to a standard curve dry chamber. Negative staining was performed by gently
created with serial dilution of synthetic Aβ oligomers and dripping 100 µl 2% aqueous uranyl acetate onto the grid,
expressed as picomolar/mg of protein. followed by removal of excess uranyl acetate solution using
a lens paper. The grids were examined in a JEOL JEM-1230
Immunoblot analysis electron microscope at 100 kV accelerating voltage. Electron
micrographs were obtained at 150,000–200,000 × magnifica-
Exosomes were prepared as described above. Brain lysates tion, for a final image scale of 3.1–4.2 pixels/nm.
were prepared from homogenised brain tissue followed by
addition of lysis buffer (150 mM NaCl, 0.5% deoxycho- Tunable resistive pulse sensing by qNano
late, 1% Triton X-100, 50  mM tris-HCL pH 7.5, 20  μl/
ml phosSTOP (Roche), 10 μl/ml Halt Protease inhibitor Exosome size and particle number were analysed by TRPS
cocktail (Thermo Fisher Scientific)), clarified by centrifu- analysis using a qNano instrument (IZON Science, UK) as
gation at 10,000 x g for 5 min and sonicated using an ultra- described previously [37]. First, isolated exosomes from
sonic probe. Cell lysates were prepared from cells collected brain tissue or conditioned media of dSH-SY5Y cells were
in lysis buffer followed by  homogenisation and  sonica- diluted and passed through a 0.2 μm filter (Millipore). Sub-
tion. Samples were mixed with 4x Laemlli loading buffer sequently, particle numbers were counted for a maximum of
and separated on a ClearPAGE SDS Gel 4–12% or 10% 5 min or until 500 particles had been counted, using 8 mbar
(C.B.S. Scientific), and transferred onto a nitrocellulose pressure and the NP150 or NP100 nanopore membranes with
membrane (Invitrogen). The gel was subsequently stained a stretch between 45 and 47 mm. Voltage was set to 0.1 and
using InstantBlue protein stain (Expedeon). Additionally, 0.25 mV to achieve a stable current. Particle size histograms
exosomes, isolated from Control and AD brains or condi- were recorded when root mean square noise was below 13
tioned media of oAβ-AF700-treated raH-SY5Y cells were pA and particle rate in time was linear. Calibration was per-
lysed by freeze-thawing and subsequently run by SEC using formed using known concentration of beads CPC70D (mode
the conditions described above. The eluted proteins were diameter 70 nm) or CPC100B (mode diameter: 110 nm) (all
collected in fractions of 1 ml, lyophilized, resuspended in from IZON) diluted in 1:500 0.2 μm filtered PBS.
15 µl PBS and spotted on 0.2 µm nitrocellulose membrane.
Membranes were then blocked by 3% BSA followed by pri- Proteinase K digestion
mary antibody incubation. The following antibodies were
used: anti-flotillin-1 (1: 500, BD Transduction Laboratories); To examine whether exosome-associated Aβ is luminal
anti-alix (1: 1000, EMD Millipore); anti-TSG101 (1:1000, or bound to the exterior exosome surface, exosomes were
Thermo Fisher Scientific); 1:1000, anti-VPS4A (Abcam), isolated from conditioned media (oAβAF700-treated) of
anti-calnexin (1:1000, Abcam), anti-synaptophysin (1:1000, dSH-SY5Y cells and incubated with proteinase K (Sigma-
Synaptic Systems), mAb158 (1: 5000/10000, BioArctic) and Aldrich,1 mg/ml) for 30 min at 37 °C. 4-(2-aminoethyl)-
anti-glyceraldehyde 3-phosphate dehydrogenase (GAPDH, benzene-sulfonyl fluoride (Sigma-Aldrich, 0.5 mM) was
1:40000, Synaptic Systems,). Anti-rabbit IgG, horseradish subsequently added to the vesicle fraction to inactivate the
peroxidase (HRP)-linked antibody (1:3000, Dako) and anti- enzyme prior to two rounds of 100,000 × g centrifugation.
mouse IgG, HRP-linked antibody (1:3000, Dako) were used The final pellet was resuspended in PBS and AF700 fluores-
as secondary antibodies. The blots were visualized using cence was measured in Tecan Safire2 microplate reader at
Amersham™ ECL™ (GE Health Care) or S ­ uperSignal® Ex/Em 696/719 nm.

13

46 Acta Neuropathologica (2018) 136:41–56

Cytotoxicity assay to the manufacturer’s instructions. GAPDH was amplified as


an internal standard. The data were calculated according to
To investigate the toxic effect of exosomes on neurons, equal the comparative Ct method to present the data as fold differ-
amounts of exosomes (based on exosomal protein estimation ences in the expression levels relative to the control sample.
by BCA) from brains or cells were added to dSH-SY5Y cells
and hiPSCs in our co-culture model for 48 h, as described
above. At the end of incubation, donor cells were removed Statistics
and cell medium was collected to assess the release of lactate
dehydrogenase (LDH) in the medium. Collected medium All statistical analyses were performed using GraphPad
was centrifuged 2000×g for 5 min at 4 °C and LDH assay Prism Software. Data were expressed as the mean ± SEM,
(Pierce) was performed according to manufacturer’s instruc- and statistical comparisons were made using two-tailed
tions. The absorbance was measured in a microplate reader unpaired Student’s t tests with Welch’s correction or one-
(SpectraMAX 190) at 490 nm with subsequent blank at way ANOVA with Tukey’s correction. Every batch of cell
680 nm. Furthermore, XTT (2,3-bis [2-methoxy-4-nitro-5- cultures was treated as one independent experiment (n = 1).
sulfophenyl]-5-[(phenylamino) carbonyl]-2H-tetrazolium P values less than or equal to 0.05 were deemed statistically
hydroxide) assay using the Cell Proliferation Kit II (Roche significant.
Diagnostics GmbH) was performed on acceptor cells accord-
ing to the manufacturer’s instructions. The reduced XTT Study approval
product produced by mitochondrial enzymes in viable cells,
formazan (bright orange in colour) was measured after 8 h The collection and use of post mortem brain tissue was
of incubation at 450 and 750 nm using a Victor 3 V 1420 approved by the Regional Ethical Committee in Uppsala,
multilabel plate reader (PerkinElmer). Both LDH and XTT Sweden (2005/103, 2005-06-29; 2009/089; 2009-04-22).
values were presented as percentage of untreated control. The process of reprogramming human cells was approved
by the Ethical Committee at Karolinska Institute, Sweden
(dnr 2012/208-31/3 with addendum 2012/856-32). A written
RNA interference informed consent was received from all donors.

Cells were seeded at a density of 12 500 cells/cm2 in a


6-well plate and transfected 24 h later with TSG101 or VPS4 Results
mRNA-targeting siRNA or a non-targeting siRNA with no
homology to any known human gene (All Stars Negative Alzheimer brain exosomes are enriched with oAβ
Control siRNA) with the HiPerFect transfection reagent (all
from Qiagen) according to manufacturer’s protocol. TSG#6 To examine whether oAβ has the potential to localize to cel-
(CAGT ​ TTA​ TCA​ TTC​ AAG​ TGT​ AA), TSG#3 (ACTG ​ TCA​ AT​ lular structures that form exosomes in the AD brain we ana-
GTT​ATT​ACT​CTA), VPS#7 (AAG​CTG​AAG​GAT​TAT​TTA​ lysed the co-localization of potential oAβ labelling and the
CGA) and VPS#5 (CTCA ​ AAG​ ACC ​ GAG​ TGA​ CAT
​ AA) siR- marker flotillin-1, expressed in multi-vesicular bodies and
NAs were used for this study. The final siRNA concentra- exosomes. Brain sections from temporal neocortex from four
tions in the culture medium ranged from 10 to 20 nmol/l. post mortem AD brains were double-immunostained with
Twenty-four hours after transfection, knockdown was veri- flotillin-1 and one of two different Aβ antibodies (mAb158
fied by quantitative real-time PCR and Western Blot analy- or 82E1). Cells with neuronal morphology displayed both
ses, and a decrease in the mRNA level of 70% or greater was mAb158 or 82E1 and flotillin-1 labelling (Fig. 1a, b). Next,
considered sufficient downregulation. the possible co-localization of mAb158 or 82E1 with flotil-
lin-1 was analysed on immunofluorescently labelled, fixed
brain sections using the same antibodies. Although the reso-
Gene expression analysis lution is not sufficient to prove co-localization, we observed
a strong correlation between mAb158 or 82E1 and flotillin-1
Total RNA was extracted with the RNeasy Mini Kit (Qia- labelling, suggesting a subcellular co-localization (Fig. 1c,
gen), and cDNA was obtained with the High Capacity d) (R2 = 0.78 and 0.86 for mAb158 and 82E1, respectively).
RNA-to-cDNA Kit (Applied Biosystems). The expression The detected fluorescence was not due to lipofuscin-derived
levels of TSG101 or VPS4 mRNA were analysed with a autofluorescence (Supplementary Fig. S1a). Currently, no
7500 Fast Real-Time PCR system and FAM/MGB probes antibodies with verified oAβ selectivity in IHC applications
(Applied Biosystems) to confirm downregulation after have been reported. Thus, we selected the two Aβ antibod-
siRNA treatment. All reactions were performed according ies with most extensive data on oligomer selectivity in other

13
Acta Neuropathologica (2018) 136:41–56 47

a c e f
Brain lysate Exosome Control AD

mAb158 flotillin-1 DAPI


mAb158 flotillin-1 DAPI

Flotillin-1 Alix

Alix Flotillin-1

Calnexin

Synaptophysin

b d g h
82E1 flotillin-1 DAPI
82E1 flotillin-1 DAPI

pM oAß / mg protein
pM oAß / mg protein
*

Control brain AD brain Control brain AD brain

i j Control brain exo


l
Control cell exo
Absorbance (215 nm)

Absorbance (700 nm)


AD brain exo
oAß cell exo
Pure Aß
Flotillin-1 pg/ml

Time (min) Time (min)

k 12 14 16 18 20 22 24 26 28 min m 12 16 18 20 22 24 26 28 min
Control brain exo Control cell exo
Control brain AD brain
AD brain exo oAß cell exo

Fig. 1  AD brain exosomes are enriched with oAβ. AD brain sections tively. Data are presented as the mean picomolar of oAβ per mg total
from temporal neocortex show co-localization of probable oAβ to exosome protein ± SEM (n = 3). *p < 0.05 by two-tailed unpaired Stu-
exosomes. a, b Double-immunostaining with the exosome marker flo- dent’s t tests with Welch’s correction. i Quantitative analysis of flo-
tillin-1 (labelled red), and two different oligomer-selective (see main tillin-1 by ELISA in control and AD brain exosomes showing equal
text) Aβ antibodies mAb158 and 82E1, respectively (labelled blue- amount of flotillin-1 between the groups. j Representative SEC pro-
green). Scale bar 10 µm. c, d Co-localization of oAβ to exosomes was file of lysed exosomes isolated from control and AD brain samples
performed after immunofluorescence labelling. mAb158 or 82E1, at 215  nm absorbance (general protein detection). k Detection of
respectively, showed substantial co-localization (yellow) with flotil- oAβ in SEC eluate fractions of AD and control brain exosomes by
lin-1 inside cells in the AD brain (R2 = 0.78 and 0.86 for mAb158 and dot blot using mAb158 antibody. l SEC chromatograms of exosomes
82E1, respectively). Scale bar, 5  µm. e Immunoblot showing flotil- isolated from conditioned media of control- or oAβ-AF700 treated
lin-1, alix, calnexin and synaptophysin in exosome and brain lysate, dSH-SY5Y cells as well as pure oAβ-AF700. Detection at 700  nm
demonstrating no cellular or synapse vesicle contamination in the absorbance (AF700 detection). m Dot blot detection of oAβ (mAb158
exosome preparation. Loading control is shown in Supplementary antibody) in SEC eluate fractions of exosomes isolated from condi-
Fig. S1d. f Immunoblot demonstrating the presence of flotillin-1 and tioned media of control- or oAβ-AF700 treated dSH-SY5Y cells of
alix, in exosome fractions isolated from control and AD brain. Quan- control and oAβ-AF700 treated dSH-SY5Y cells exosomes. The anal-
titative ELISA analysis of oAβ in exosomes isolated from AD and ysis of SEC fractions confirms the presence of oAβ in exosomes
control brains, using 82E1 (h) and mAb158 (g) antibodies, respec-

assays (mainly ELISA). Both these antibodies showed including multivesicular bodies, in the human AD brain,
similar labelling patterns in brain tissue (Fig. 1a–d). Fur- which thus suggests that oAß could be released in exosomes.
thermore, our findings from dot-blot (Fig. 1k, m, described Since oAβ is believed to be central to AD pathophysi-
below) supports that mAb158 has a selectivity for aggre- ology [50], we postulated that exosomes from AD brains
gates over monomers also in assays where the protein is would contain higher concentrations of oAβ when com-
in a bound state. Taken together, these data indicate that pared to similar preparations of control brain samples from
the identified Aβ labelling likely represents oligomeric Aβ patients deceased from non-neurological reasons. Using
aggregates. Thus, our findings suggest an association of well-established methods for mild dissociation of nerv-
oAβ with intracellular compartments containing flotillin-1, ous tissue followed by separation of extracellular matrix
from cells we were able to isolate exosomes by sequential

13

48 Acta Neuropathologica (2018) 136:41–56

Exosomes oAß
a b c

Or

labeling of exosomes
containing oAβ from
AD brain or cells in culture
d e
2. Uptake by donor cells
on glass slides

figure b-c
and
figure f-g

3. Co-culture slides upside down


on top of acceptor cells

f g

4. Co-culture for 48 h

5. Removal of the donor cell slide


Merged
Red

figure d-e
and
figure h

confocal microscopy and cytotoxicity


assays

i j
Exosomes PKH67 Exosome free fraction PKH67 CD63 GFP expressing exosomes Exosomes PKH26

13
Acta Neuropathologica (2018) 136:41–56 49

◂Fig. 2  Exosome-mediated uptake and propagation of oAβ in neu- free oAß during the extraction as spiking exosomes with
ronal cells. Exosomes isolated from brain tissue or conditioned exogenous oAß did not result in increased levels, which
media of dSH-SY5Y cells were labelled with the dye PKH67 and
added to donor hiPSCs or dSH-SY5Y cells. After 3  h of incubation
also implies that association of oAß with exosomes occurs
at 37  °C, donor cells were fixed, stained with mAb158 (for brain intracellularly (Supplementary Fig. S1e). Additionally, the
exosomes) and analysed by confocal microscopy or donor cells were presence of oAβ in isolated exosomes was further assessed
co-cultured with another set of hiPSCs or dSH-SY5Y (recipient using size exclusion chromatography (SEC). SEC chroma-
cells). After 48 h of co-culture, donor cells were removed and recipi-
ent cells were fixed, stained with mAb158 (for brain exosomes) and
tograms of the isolated and lysed exosomes from control
analysed by confocal microscopy. a A cartoon illustrating the co- and AD brain samples were analysed at 215 nm absorbance
culture model with hiPSC or dSH-SY5Y cells employed to measure (general protein detection) and proteins were detected in the
the transfer of the brain or cell exosomes containing oAβ. Uptake of void volume (first peak) as well as in fractions correspond-
b control and c AD brain exosomes (green) containing oAβ (red) in
hiPSC donor  cells. Transfer of AD brain exosomes (green) contain-
ing to lower molecular weight (Fig. 1j). The presence of
ing oAβ (red) to recipient d hiPSCs and e dSH-SY5Y cells. Uptake oAβ in the different SEC eluate fractions was further inves-
of exosomes (green) containing oAβ-AF700 (red) in donor f dSH- tigated by dot-blot analysis using mAb158. This confirmed
SY5Y and g hiPSCs. h Transfer of oAβ-AF700 containing exosomes that exosomes isolated from AD brain samples contained
in recipient dSH-SY5Y cells. Super-imposed image of the red (oAβ)
and green (exosomes) channels on a DIC image shows co-localization
oAβ in SEC fractions running at the oligomer size that
(yellow) of exosomes and oAβ. Arrows indicate exosomes or exo- was not detected in control brain (Fig. 1k). Similarly, lysed
some containing oAβ. i Cellular uptake of isolated brain exosomes exosomes isolated from conditioned media of control- or
and brain exosome free fraction after PKH67 staining showing no oAβ-AF700 treated neuronally  differentiated neuroblas-
PKH67 uptake in the absence of exosomes. j Isolated exosomes from
CD63-GFP expressing SH-SY5Y cells, double-labelled with PKH26
toma cells (dSH-SY5Y) as well as pure oAβ-AF700 were
(red), were added to dSH-SY5Y cells. The CD63-GFP were intensi- analysed on SEC chromatograms with detection at 700 nm
fied using an anti-GFP antibody. Scale bar (b–h), j 20 µm, i 10 µm absorbance (AF700 detection). This shows absorbance
peaks (oligomeric and monomeric) at the same time points
(size) for exosomes from conditioned media of oAβ-AF700
ultracentrifugation from fresh-frozen, post mortem brain tis- treated cells as for the pure oAβ-AF700 initially used to treat
sues from temporal neocortex of both AD and control sub- the cells (Fig. 1l). oAβ was detected only in fractions from
jects. Analyses of the isolated fractions by Tunable Resis- exosomes from conditioned media of oAβ-AF700 treated
tive Pulse Sensing technology (TRPS, qNano system) and dSH-SY5Y cells and not in control cell exosomes (Fig. 1m).
transmission electron microscopy (TEM, Supplementary Furthermore, these findings show that, in addition to previ-
Fig. S1b, c) demonstrated presence of vesicles with a size ously shown Aβ oligomer selectivity in ELISA assay [19,
corresponding to exosomes [55]. Immunoblotting confirmed 57], the mAb158 antibody is oligomer selective also in an
the extracellular origin of the extracted vesicles by presence assay where protein is not free floating.
of the exosomal markers alix and flotillin-1 and the lack of The localization of oAß to exosomes was further inves-
the cellular marker calnexin and the synapse vesicle marker tigated in neuronal cell models. dSH-SY5Y or hiPSC were
synaptophysin (Fig. 1e). Furthermore, immunoblotting con- incubated with oAβ-AF700 for 3 h, after 48 h exosomes were
firms the presence of exosomal markers in extracted vesi- isolated from the conditioned media by sequential ultracen-
cles from both control and AD brains (Fig. 1f). As immu- trifugation. Similar to the brain extractions, the isolated vesi-
nostaining of flotillin-1 and oAβ in the AD brain sections cles were characterized as exosomes using immunoblotting
does not have the sufficient resolution, ELISA, which is for the exosome markers, alix and flotillin-1, lack of cellular
a more quantitative method, was implemented to demon- marker calnexin and synapse vesicle marker synaptophysin,
strate the oAβ in brain exosomes. When used as capture and TRPS and TEM (Supplementary Fig. S2a–d) [35, 54]. To
target in an ELISA the Aβ antibodies mAb158 is selective investigate the location of oAß, exosomes isolated from con-
for Aβ oligomers [19, 57], while 82E1 detects both soluble ditioned media from oAß-AF700 treated retinoic acid dif-
and fibril Aβ [31]. Interestingly, ELISA with either one of ferentiated SH-SY5Y cells (raSH-SY5Y) were treated with
these two oligomer-selective antibodies showed significantly proteinase K (1 mg/ml) to digest proteins on the surface,
higher levels of oAß in exosomes from AD brains (n = 5, confirming that the localization of oAβ is mainly luminal
3.12 ± 0.93 pM oAß/mg protein for 82E1 and 7.69 ± 1.68 pM as this did not significantly decrease exosomal oAβAF700
oAß/mg protein for mAb158) than in those from healthy (Supplementary Fig. S2e).
control brains (n = 5, 0.46 ± 0.30 pM oAß/mg protein for
82E1 and 2.66 ± 0.74 pM oAß/mg protein for mAb158) Exosomes from AD brain are internalized
(Fig.  1g, h). There was  no significant difference in the and transferred by neurons, causing cytotoxicity
amount of flotillin-1 in exosomes derived from AD or con-
trol brain tissue of equivalent weight (Fig. 1i). The increase To investigate whether exosomes could be a vehicle for the
in exosomal oAß was not an effect of co-precipitation of spreading of oAß, we investigated if exosomes isolated from

13

50 Acta Neuropathologica (2018) 136:41–56

AD brains could be taken up by hiPSCs and dSH-SY5Y cells Further evidence for the exosomal transportation of oAβ
(Fig. 2a). The cells were incubated with exosomes (PKH67 was obtained by using a transwell co-culture model after
labelled) isolated from brain tissue. After 3 h, both exosomes uptake of oAβ containing exosomes to donor cells. Also,
(Fig. 2b) and oAß (mAb158, Fig. 2c) had been taken up by in this model oAβ transferred to recipient cells, suggesting
the cells. Using a co-culture system with hiPSCs or dSH- vesicle transfer of oAβ between cells without direct neuritic
SY5Y cells, previously shown to result in synaptically con- connections. However, compared to the coverslip co-culture
nected neurons [40], we next investigated the possible trans- model significantly less neurons containing transfer of oAβ
fer of brain-derived exosomal oAß between these neurons. was detected in the transwell model, a decrease from 23.60
When RFP labelled donor cells that had been exposed to to 5.18% as analysed by flow cytometry and illustrated by
exosomes were placed on top of recipient neurons for 48 h, confocal imaging (Supplementary Fig. S4a, b). Thus, sug-
a substantial number of transferred exosomes (PKH67) and gesting that transfer is more efficient between cells in prox-
oAβ (mAb158) could be detected in both types of recipi- imity, which could potentially depend on multiple mecha-
ent cells (Fig. 2d, e). Moreover, transferred oAß was still nisms of transfer.
partly co-localized with PKH67 labelled exosomes in the Next, we investigated if the spread of exosomes and their
recipient cells, suggesting that after being internalized part oAß cargo could result in neuronal toxicity. Isolated brain
of the exosomes can be transferred onwards, still intact with exosomes (equalized by protein amounts) from either control
their oAß content. Similarly, uptake of oAß in raSH-SY5Y or AD brains were added to dSH-SY5Y cells for 3 h, these
treated with oAβ-AF700 (3 h) resulted in a co-localization cells were used as donor cells in 48 h co-culture with recipi-
with intracellular flotillin-1 and TSG101 (Supplementary ent dSH-SY5Y cells. The recipient cells were subsequently
Fig. S3a-b). Isolated (48 h conditioned media) and labelled investigated for morphological changes compared to control
(PKH67) exosomes were readily taken up by new cells. A co-culture with untreated donor cells (Fig. 3a). As described
high degree of internalization of exosomes in either dSH- previously [2] the control dSH-SY5Y shows neuronal mor-
SY5Y (Fig. 2f) or hiPSC (Fig. 2g) cells was observed. These phology with long, branching networks of neurites. Similar
vesicular structures had a mainly perinuclear localization morphology was seen in recipient cells in the control brain
and many of them were positive for oAβ-AF700. Using exosome conditions. Occasional changes in neurite mor-
these secondary neurons as donor cells and co-culturing phology could be identified. Interestingly, more pronounced
them with a new set of dSH-SY5Y recipient cells for 24 h pathology was seen in recipient cells co-cultured with AD
showed further spread of AF700 labelled oAβ to this third brain exosome treated donor cells. Here dystrophic neur-
set of cells (red channel, Fig. 2h). Similarly to what we ites with neurite beading and loss of neurite branching were
observed for exosomes from AD brains, we could detect a identified, known early signs of neurodegeneration [51]. We
substantial fraction of oAβ that was still co-localized with quantified these effects further and notably, we found that
exosomes (merged, Fig. 2h). These findings support that, the transfer of AD brain exosomes induced significant cyto-
apart from releasing their cargo, intact exosomes also can toxicity compared to control brain exosomes, as assessed by
carry and transfer oAβ further to new recipient cells. Like- the degree of membrane leakage of lactate dehydrogenase
wise, exosomes isolated from conditioned media from cells enzyme (LDH) from the recipient hiPSC and dSH-SY5Y
that had not been exposed to oAβ were also taken up by cells (Fig. 3b, c). Induced cellular toxicity was also detected
cells and transferred onwards (Supplementary Fig. S3c), by XTT (2,3-bis [2-methoxy-4-nitro-5-sulfophenyl]-5-
corroborating that the release and uptake of exosomes per [(phenylamino) carbonyl]-2H-tetrazolium hydroxide) analy-
se is not dependent on the presence of oAß. To confirm that sis in both cell types (Fig. 3d, e). Similar toxicity was further
the observed PKH labelling is not due to artefacts we used seen after treatment with exosomes from conditioned oAß
the supernatant from the last ultracentrifugation wash-step media (Fig. 5d). In addition, exosomes from control brains
of the exosome isolation from brain or conditioned media, which should contain significantly less oAß, caused toxicity
respectively. The supernatant was labelled with PKH67 in hiPSCs but not in dSH-SY5Y cells (Fig. 3b, e). No toxic-
using the same protocol as for labelling the exosomes. The ity was seen after treatment with exosomes from conditioned
labelled sample were subsequently added to raSH-SY5Y media from cells not treated with oAβ (Supplementary Fig.
cells. Contrary to the exosome fraction the exosome-free S5a). Taken together, these results suggest that intact AD
fraction did not result in any labelling in the cells either in brain exosomes, carrying oAβ, are taken up by neurons and
brain (Fig. 2i) or conditioned media extracts (Supplemen- migrate to second order neurons where they can release their
tary Fig. S3d). Furthermore, isolated exosomes from CD63- cargo and cause cytotoxicity.
GFP expressing raSH-SY5Y cells were double-labelled with
PKH26 (red) and added to raSH-SY5Y cells (Fig. 2j). This
confirmed that the exosomes studied here are not artefacts,
but indeed exosomes.

13
Acta Neuropathologica (2018) 136:41–56 51

a Control Control brain exo AD brain exo

hiPSC dSH-SY5Y
b c *
*
***

Cell viability LDH (%)


Cell viability LDH (%)

***
ns

** -------------------------------

-------------------------------

AD brain Control brain AD brain Control brain

d hiPSC e dSH-SY5Y
ns
Cell viability XTT (%)

Cell viability XTT (%)


-------------------------------
ns
ns
-------------------------------
*** **
**

AD brain Control brain AD brain Control brain

Fig. 3  Transfer of AD brain exosomes causes cytotoxicity. Exosomes shown in magnified insert. The conditioned media was collected for
isolated from control and AD brain tissues were added to donor hiP- LDH assay (b, c) and recipient cell viability evaluated by XTT (d, e).
SCs or dSH-SY5Y cells. After 3 h of incubation at 37 °C, donor cells Values are expressed as percentage of untreated control. Values are
were washed with PBS and co-cultured with another set of hiPSCs mean ± SEM (n = 6 separate experiments). LDH shows that transfer
or dSH-SY5Y (recipient cells). After 48 h of co-culture, donor cells of AD brain exosomes causes significant higher cytotoxicity com-
were removed. a Morphological changes assessed in recipient dSH- pared to control brain exosomes in both cell types. NS, not signifi-
SY5Y showing loss of neurite branching after transfer of AD brain cant; *p < 0.05, **p < 0.01, ***p < 0.001 by two-tailed unpaired Stu-
exosomes. Also, neurite beading was seen in dystrophic neurites as dent’s t tests with Welch’s correction

Inhibition of exosome formation and secretion VPS4A) (Fig. 4a–c) was achieved. The siRNA treatment
inhibit the spread of oAβ did not affect viability, as analyses showed that > 95% of
the cells were viable as measured by XTT assay (Fig. 4d).
If exosomes are capable of transferring oAß between neu- To distinguish between different mechanisms of transfer, we
rons, it should be possible to stop this transfer by inhibiting again used the coverslip and transwell co-culture models
the formation of exosomes. As previously shown, biogen- and quantified the oAβ transfer in presence of TSG101 or
esis of exosomes and its cargo proteins can be modulated VPS4A siRNA in our co-culture model using flow-cytome-
by knocking down two Endosomal Sorting Complexes try. Interestingly, upon inhibition of exosome formation and
Required for Transport (ESCRT) proteins, TSG101 and secretion the oAβ transfer was almost completely blocked in
VPS4A, required for exosome formation and secretion, the transwell model, whereas almost half of the oAβ transfer
respectively [3, 18, 21, 30]. By using siRNA oligonucleo- could be stopped in the coverslip model (Fig. 4e). These
tides, a significant decrease in the mRNA levels (decreased results indicate that exosomes are largely responsible for the
in average with 90% for TSG101 and 88% for VPS4A), neuron-to-neuron transfer of oAβ, although other mecha-
protein levels (decreased in average with 87% for TSG101 nisms of transfer may also be involved.
and 65% for VPS4A) and the number of secreted exosomes
(decreased in average with 95% for TSG101 and 76% for

13

52 Acta Neuropathologica (2018) 136:41–56

a b
TSG101 VPS4A
Relative mRNA expression

Relative protein levels


-------------------------------------------

46 kDa 50 kDa
*

*** *** **
GAPDH

TSG101 VPS4A siRNA Neg C TSG101 Neg C VPS4A Neg C TSG101 VPS4A

c d e
Number of exosomes (%)

Cell viability XTT (%)


------------------------------------------ -------------------------------------------

oAß transfer (%)


----------------------------------------- TSG101
*** VPS4A
***
**

*** *** ***

TSG101 VPS4A Neg C TSG101 VPS4A Coverslip Transwell

Fig. 4  Downregulation of exosomal proteins TSG101 and VPS4A toxicity was detected by XTT assay after 48  h of transfection with
inhibits the spread of oAβ. Depletion of TSG101 and VPS4A by siRNA. e Quantification of oAβ transfer in presence of TSG101 or
siRNA in dSH-SY5Y cells. a Real time PCR analysis of mRNA VPS4A siRNA in both coverslip and transwell co-culture model by
expressions to show knock down efficiency of transfected cells by flow cytometry, shows that both siRNAs significantly inhibit oAβ
TSG101 or VPS4A siRNA. b Representative immunoblot picture of transfer. Values are expressed as percentage of siRNA negative con-
cell lysates after siRNA treatment for 72  h and associated densito- trol and indicated as dotted line. Values are the mean ± SEM (n = 4
metric analysis (n = 3). c Bead flow cytometry analysis of exosomes separate cultures), *p < 0.05, **p < 0.01, ***p < 0.001 by two-tailed
shows a significant decrease in the number of secreted exosomes after unpaired Student’s t tests with Welch’s correction
TSG101 or VPS4A siRNA treatment in raSH-SY5Y cells. d No cyto-

The uptake of exosomes and spread of oAß with dynasore treatment, which almost completely abolished
is dynamin‑dependent and can be blocked the uptake of exosomes (Fig. 5a), while having only minor
effect on the internalization of oAβ added directly to the
After being released, the exosomes can be taken up by the medium (Fig. 5b). This observation supports that the uptake
recipient cells via different mechanisms. In dSH-SY5Y of exosomes, and thus also oAß transferred via this route, is
cells the uptake of isolated exosomes observed at 37 °C, as regulated by dynamin.
described above, was completely abrogated at 4 °C (Supple- The dependence on dynamin, and thus exosomes, for
mentary Fig. S5b). Thus, the exosomal uptake is an active the spread of oAß between neurons was further confirmed
process. Neuronal cells are capable of different modes of using dSH-SY5Y cells in the transwell model using donor
endocytosis, ranging from receptor-mediated and clathrin- cells fed with exosomes, isolated from conditioned media
dependent, to independent endocytosis [10, 34, 47]. To (oAβ-AF700 treated raSH-SY5Y cells). In this setting with
explore the uptake mechanism, we treated dSH-SY5Y cells an absence of direct neuritic connections, oAβ transfer was
with isolated, PKH67 labelled, exosomes together with vari- almost completely blocked by dynamin inhibition (Fig. 5c),
ous inhibitors of endocytosis: dynasore (dynamin inhibitor), in line with a major dependence on exosomes for transfer.
phenylarsine oxide (clathrin inhibitor) and genistein (cave- When instead performing the same experiment using the
olae inhibitor). Treatment with phenylarsine oxide caused coverslip co-culture model, allowing for direct cell-to-cell
minor cellular toxicity, while no notable cellular toxicity was contacts, there was also a significant decrease of transferred
observed upon treatment with dynasore or genistein (Sup- oAß (Fig.  5c). Since our observations substantiate the
plementary Fig. S5c). Strikingly, all three inhibitors caused hypothesis that exosomes play a significant role in trans-
a significant decrease in the proportion of cells that took ferring oAβ from one neuron to another, we next sought
up exosomes as quantified by flow-cytometry; the decrease to investigate whether transfer of exosomes containing oAβ
was 97.7% with dynasore, 66.5% with phenylarsine oxide had any direct toxic effect on the recipient cells. Cell-to-
and 76.0% with genistein. Thus, the effect was most marked cell transfer of exosomes, isolated from conditioned media

13
Acta Neuropathologica (2018) 136:41–56 53

a b
--------------------------------------------- ns

Exosome uptake (%)

oAß uptake (%)


- - - - - - - - - - - - - - - - - - - - - - - -ns
--------------------------
**

***
***

***

Dynasore Phenylarsine Genistein Dynasore Phenylarsine Genistein


oxide oxide

c d **
**
-----------------------------------------------

Cell viability LDH (%)


oAß transfer (%)

- - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -ns
-----------

ns

***
***

Coverslip Transwell oAß oAß+Dynasore

Fig. 5  The uptake of exosomes and the subsequent spreading of oAß nificant decrease of the proportion of cells with oAβ transfer in both
is dynamin-dependent. a, b Uptake of PKH67 labelled exosomes models (control, dotted line). d Transfer of exosomes isolated from
or oAβ-AF700 in dSH-SY5Y cells. Cells were pre-incubated with oAβ treated cells causes cytotoxicity in recipient cells compared to
the indicated inhibitors for 30  min, then exposed to exosomes or untreated control as shown by LDH assay, whereas dynasore treat-
oAβ-AF700. After 3  h incubation, samples were collected and ment significantly reduces the cytotoxic effect versus untreated con-
the proportion of cells with uptake was quantified by flow cytome- trol (dotted line). Data are represented as the mean ± SEM, NS, not
try and related to untreated control (dotted line). c Flow cytometry significant; n = 4; **p < 0.01, ***p < 0.001 by two-tailed unpaired
analysis of oAβ transfer in presence of dynasore in coverslip and Student’s t tests with Welch’s correction
transwell co-culture models. After dynasore treatment there is a sig-

(oAβ-AF700 treated raSH-SY5Y cells), showed a significant cell cultures [46] have been shown to contain monomeric
increase in cellular toxicity of 26% in the recipient cell, as Aβ and tau, but so far, no study has addressed the presence
evident by LDH assay (Fig. 5d). Exosomes from untreated of oAβ in exosomes from human AD brains. In this study,
raSH-SY5Y cells had no significant toxic effect on the recip- we could show that AD brain exosomes contain an increased
ient cells (Supplementary Fig. S5a). Importantly, we found amount of oAβ compared to non-neurological control brains
that such induced toxicity could be abolished by blocking the and found evidence that exosomes can be responsible for
transfer of exosomes using dynasore (Fig. 5d). These data the neuron-to-neuron transfer of toxic oAβ. These findings
provide proof-of-concept that the transfer of exosomes and suggest that exosomes might be the main mediator of the
oAβ together with subsequent toxicity, can be prevented by pathogenic progression in AD as was recently suggested for
inhibiting the dynamin-dependent uptake pathway. dementia with Lewy bodies [41]. In addition, we found a
co-localization between oAβ and exosomes inside neurons,
which might indicate that exosomes play a role in Aβ sort-
Discussion ing and oligomerization [16]. The AD brain exosomes were
further shown to effectively transfer oAβ from one neuron
Recent evidence suggests that toxic Aβ aggregates can to another, with subsequent toxic effects on the recipient
spread pathology in the Alzheimer brain. The nature of the cells. Interestingly, at least a part of the exosomes seems
propagating species has not been established, although sev- to be transferred intact to further cells, consistent with a
eral studies have indicated a particularly pathogenic role of recent study showing that a substantial fraction of exosomes
soluble oAβ on synaptic and cellular functions and structure internalized in one cell were subsequently passed on to a
[13, 32, 50, 60]. It has been speculated that exosomes might second cell [44]. Importantly, there is increasing evidence of
transfer neurodegenerative proteins in the affected brain [28]. correlations between intra-neuronal oAβ and cell death [43].
Accordingly, exosomes from blood [22], CSF [17, 49] and We have previously demonstrated that transfer of oAβ causes

13

54 Acta Neuropathologica (2018) 136:41–56

neurotoxicity [40] which has also been shown with Aß con- spread and toxicity of oAβ may lead to the identification of
taining exosomes isolated from AD CSF [33]. Accordingly, new pharmaceutical targets for AD.
we now found signs of neurotoxicity both morphologically
and with the LDH and the XTT assays after transfer of Acknowledgements  The authors like to thank Dr. Anna Falk for the
kind gift of the AF22 cells, MSc. Chris Sackmann for the kind gift
exosomes carrying oAβ. This observation not only rein- of the CD63-GFP SH-SY5Y cells and Dr. Jakob Domert for expert
forces the role of intracellular oAβ in AD pathogenesis but illustrations. Xandra Breakefield is acknowledged for critical proof-
also establishes the disease relevance associated with the reading of the manuscript. This research was made possible by fund-
exosomal neuron-to-neuron transfer of intercellular oAβ. ing from the Swedish Research Council (MH: 523-2013-2735), The
Swedish Alzheimer foundation, The Swedish Brain Foundation, the
The molecular content of exosomes is a fingerprint of Hans-Gabriel and Alice Trolle-Wachtmeister Foundation for Medical
the releasing cell type and, because of their small size, Research, Konung Gustaf V:s och Drottning Victorias Frimurarestif-
neuronal exosomes are released into accessible body fluids telse, Marianne and Marcus Wallenberg Foundation, The Swedish
such as blood and CSF [48]. Since neuronal exosomes dis- Fund for Research without Animal Experiments, The Swedish Demen-
tia Foundation, the Linköping University Neurobiology Centre and
play unique neuron-specific surface markers [22, 26] they the County Council of Östergötland. The funders had no role in study
may be a valuable biomedical marker for early diagnosis design, data collection and analysis, decision to publish, or preparation
and treatment in AD. Indeed, exosomes have recently been of the manuscript.
highlighted as diagnostic biomarkers in various disease con-
ditions, including AD [29, 36]. In concordance, the finding Author contributions  MSS and AA designed the experimental
approach, performed all the major biochemical and cellular experi-
of increased levels of oAβ in brain exosomes opens the pos- ments, analysed and interpreted the data, generated the figures, partici-
sibility that similar features could be detected also in eas- pated in the study design, and in writing the manuscript. LC carried
ily accessible body fluids, such as plasma and CSF. Hence, out SEC and DOT blot, prepared the Aβ aggregates and participated
measurement of increased oAβ in exosomes from such in writing the manuscript. CH prepared brain slices and carried out
immunohistochemistry and participated in writing the manuscript.
patient samples could potentially serve as a diagnostic tool. ML carried out EM and participated in writing the manuscript. LA
The intercellular propagation of oAβ and its ensuing provided antibodies and participated in writing the manuscript. MI
toxicity could also serve as a potential treatment target by provided brain samples and participated in writing the manuscript. MH
inhibiting either formation, secretion, or cellular uptake of conceived the hypothesis, coordinated, and led the study, participated
in study design, data interpretation and in writing the manuscript.
exosomes. Indeed, downregulation of TSG101 and VPS4A,
proteins necessary for exosome formation and secretion,
was found to result in decreased release of exosomes and
Compliance with ethical standards 
a reduced subsequent transfer of oAβ, thus supporting the Conflict of interest  The authors have declared that no conflict of inter-
possibility of modulating this mechanism. Moreover, these est exists.
observations are in line with recent studies showing that
interfering with exosome release can impact the release of Open Access  This article is distributed under the terms of the Crea-
specific proteins [9, 14]. An alternative therapeutic target tive Commons Attribution 4.0 International License (http://creat​iveco​
mmons​.org/licen​ses/by/4.0/), which permits unrestricted use, distribu-
could be the dynamin-dependent uptake of exosomes [23, tion, and reproduction in any medium, provided you give appropriate
24] as the dynamin inhibitor dynasore decreased exosome credit to the original author(s) and the source, provide a link to the
propagation, spread of oAβ and the associated neuronal tox- Creative Commons license, and indicate if changes were made.
icity, leading to rescued cell viability. Dynasore itself would
not be a feasible therapeutic substance, but phenothiazine-
derived antipsychotic drugs have been suggested to inhibit References
dynamin dependent endocytosis [11] and could thus be suit-
able for further drug development. 1. Agholme L, Lindstrom T, Kagedal K, Marcusson J, Hallbeck M
In conclusion, our results point to a role for exosomes (2010) An in vitro model for neuroscience: differentiation of SH-
in the spreading of toxic oAβ and the associated disease SY5Y cells into cells with morphological and biochemical char-
acteristics of mature neurons. J Alzheimers Dis 20:1069–1082.
progression in the AD brain (summarized in Supplemen- https​://doi.org/10.3233/JAD-2010-09136​3
tary Fig. S6). It has been suggested that exosomal release 2. Agholme L, Nath S, Domert J, Marcusson J, Kagedal K, Hallbeck
may provide an alternative disposal mechanism to lysoso- M (2013) Proteasome inhibition induces stress kinase dependent
mal degradation of oAβ [5] or other proteins that are resist- transport deficits—implications for Alzheimer’s disease. Mol Cell
Neurosci 58C:29–39. https​://doi.org/10.1016/j.mcn.2013.11.001
ant to degradation [55]. We speculate that this alternative 3. Akrap I, Thavamani A, Nordheim A (2016) Vps4A-mediated
mechanism of clearance, which initially could be beneficial tumor suppression upon exosome modulation? Ann Transl Med
for the cells, over time becomes a liability with increased 4:180. https​://doi.org/10.21037​/atm.2016.04.18
propagation of pathological proteins throughout the brain. 4. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T,
Wolozin B, Butovsky O, Kügler S, Ikezu T (2015) Depletion of
The possibility of inhibiting exosome transfer and the related microglia and inhibition of exosome synthesis halt tau propaga-
tion. Nat Neurosci. https​://doi.org/10.1038/nn.4132

13
Acta Neuropathologica (2018) 136:41–56 55

5. Bellingham SA, Guo BB, Coleman BM, Hill AF (2012) PLoS One 7:e29597. https​://doi.org/10.1371/journ​al.pone.00295​
Exosomes: vehicles for the transfer of toxic proteins associated 97
with neurodegenerative diseases? Front Physiol 3:124. https:​ //doi. 21. Fevrier B, Raposo G (2004) Exosomes: endosomal-derived vesi-
org/10.3389/fphys​.2012.00124​ cles shipping extracellular messages. Curr Opin Cell Biol 16:415–
6. Blockhuys S, Celauro E, Hildesjo C, Feizi A, Stal O, Fierro-Gon- 421. https​://doi.org/10.1016/j.ceb.2004.06.003
zalez JC, Wittung-Stafshede P (2017) Defining the human cop- 22. Fiandaca MS, Kapogiannis D, Mapstone M, Boxer A, Eitan E,
per proteome and analysis of its expression variation in cancers. Schwartz JB, Abner EL, Petersen RC, Federoff HJ, Miller BL
Metallomics 9:112–123. https​://doi.org/10.1039/c6mt0​0202a​ et al (2015) Identification of preclinical Alzheimer’s disease by a
7. Braak H, Braak E (1991) Neuropathological staging of Alzheimer- profile of pathogenic proteins in neurally derived blood exosomes:
related changes. Acta Neuropathol 82:239–259 a case-control study. Alzheimers Dement 11(600–607):e601. https​
8. Brettschneider J, Del Tredici K, Lee VM, Trojanowski JQ (2015) ://doi.org/10.1016/j.jalz.2014.06.008
Spreading of pathology in neurodegenerative diseases: a focus 23. Fitzner D, Schnaars M, van Rossum D, Krishnamoorthy G,
on human studies. Nat Rev Neurosci 16:109–120. https​://doi. Dibaj P, Bakhti M, Regen T, Hanisch UK, Simons M (2011)
org/10.1038/nrn38​87 Selective transfer of exosomes from oligodendrocytes to micro-
9. Bulloj A, Leal MC, Xu H, Castano EM, Morelli L (2010) Insulin- glia by macropinocytosis. J Cell Sci 124:447–458. https​://doi.
degrading enzyme sorting in exosomes: a secretory pathway for org/10.1242/jcs.07408​8
a key brain amyloid-beta degrading protease. J Alzheimers Dis 24. Fruhbeis C, Frohlich D, Kuo WP, Amphornrat J, Thilemann S,
19:79–95. https​://doi.org/10.3233/JAD-2010-1206 Saab AS, Kirchhoff F, Mobius W, Goebbels S, Nave KA et al
10. Cosker KE, Segal RA (2014) Neuronal signaling through endo- (2013) Neurotransmitter-triggered transfer of exosomes mediates
cytosis. Cold Spring Harb Perspect Biol. https​://doi.org/10.1101/ oligodendrocyte-neuron communication. PLoS Biol 11:e1001604.
cshpe​rspec​t.a0206​69 https​://doi.org/10.1371/journ​al.pbio.10016​04
11. Daniel JA, Chau N, Abdel-Hamid MK, Hu L, von Kleist L, Whit- 25. Fu H, Hussaini SA, Wegmann S, Profaci C, Daniels JD, Herman
ing A, Krishnan S, Maamary P, Joseph SR, Simpson F et al (2015) M, Emrani S, Figueroa HY, Hyman BT, Davies P et al (2016) 3D
Phenothiazine-derived antipsychotic drugs inhibit dynamin and Visualization of the temporal and spatial spread of tau pathology
clathrin-mediated endocytosis. Traffic 16:635–654. https​://doi. reveals extensive sites of tau accumulation associated with neu-
org/10.1111/tra.12272​ ronal loss and recognition memory deficit in aged tau transgenic
12. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow mice. PLoS One. https​://doi.org/10.1371/journ​al.pone.01594​63
AR, Zhu L, Vanderburg CR, McLean PJ (2012) Exosomal cell-to- 26. Goetzl EJ, Boxer A, Schwartz JB, Abner EL, Petersen RC, Miller
cell transmission of alpha synuclein oligomers. Mol Neurodegener BL, Kapogiannis D (2015) Altered lysosomal proteins in neu-
7:42. https​://doi.org/10.1186/1750-1326-7-42 ral-derived plasma exosomes in preclinical Alzheimer disease.
13. DeKosky ST, Scheff SW (1990) Synapse loss in frontal cortex Neurology 85:40–47. https​://doi.org/10.1212/WNL.00000​00000​
biopsies in Alzheimer’s disease: correlation with cognitive sever- 00170​2
ity. Ann Neurol 27:457–464. https​://doi.org/10.1002/ana.41027​ 27. Gouras G, Tampellini D, Takahashi R, Capetillo-Zarate E (2010)
0502 Intraneuronal beta-amyloid accumulation and synapse pathology
14. Dinkins MB, Dasgupta S, Wang G, Zhu G, Bieberich E (2014) in Alzheimer’s disease. Acta Neuropathol 119:523–541. https​://
Exosome reduction in vivo is associated with lower amyloid doi.org/10.1007/s0040​1-010-0679-9
plaque load in the 5XFAD mouse model of Alzheimer’s disease. 28. Guo JL, Lee VM (2014) Cell-to-cell transmission of pathogenic
Neurobiol Aging 35:1792–1800. https​://doi.org/10.1016/j.neuro​ proteins in neurodegenerative diseases. Nat Med 20:130–138.
biola​ging.2014.02.012 https​://doi.org/10.1038/nm.3457
15. Domert J, Rao SB, Agholme L, Brorsson AC, Marcusson J, 29. Hamlett ED, Goetzl EJ, Ledreux A, Vasilevko V, Boger HA,
Hallbeck M, Nath S (2014) Spreading of amyloid-beta pep- LaRosa A, Clark D, Carroll SL, Carmona-Iragui M, Fortea J et al
tides via neuritic cell-to-cell transfer is dependent on insufficient (2016) Neuronal exosomes reveal Alzheimer’s disease biomarkers
cellular clearance. Neurobiol Dis. https​: //doi.org/10.1016/j. in Down syndrome. Alzheimers Dement. https:​ //doi.org/10.1016/j.
nbd.2013.12.019 jalz.2016.08.012
16. Edgar JR, Willen K, Gouras GK, Futter CE (2015) ESCRTs 30. Hasegawa T, Konno M, Baba T, Sugeno N, Kikuchi A, Kobayashi
regulate amyloid precursor protein sorting in multivesicular M, Miura E, Tanaka N, Tamai K, Furukawa K et al (2011) The
bodies and intracellular amyloid-beta accumulation. J Cell Sci AAA-ATPase VPS4 regulates extracellular secretion and lysoso-
128:2520–2528. https​://doi.org/10.1242/jcs.17023​3 mal targeting of alpha-synuclein. PLoS One 6:e29460. https:​ //doi.
17. Eitan E, Hutchison ER, Marosi K, Comotto J, Mustapic M, org/10.1371/journ​al.pone.00294​60
Nigam SM, Suire C, Maharana C, Jicha GA, Liu D et al (2016) 31. Horikoshi Y, Sakaguchi G, Becker AG, Gray AJ, Duff K, Aisen
Extracellular vesicle-associated abeta mediates trans-neuronal PS, Yamaguchi H, Maeda M, Kinoshita N, Matsuoka Y (2004)
bioenergetic and Ca2 + -handling deficits in alzheimer’s disease Development of Abeta terminal end-specific antibodies and sen-
models. NPJ Aging Mech Dis. https​://doi.org/10.1038/npjam​ sitive ELISA for Abeta variant. Biochem Biophys Res Commun
d.2016.19 319:733–737. https​://doi.org/10.1016/j.bbrc.2004.05.051
18. Eitan E, Suire C, Zhang S, Mattson MP (2016) Impact of lyso- 32. Hsia AY, Masliah E, McConlogue L, Yu GQ, Tatsuno G, Hu K,
some status on extracellular vesicle content and release. Ageing Kholodenko D, Malenka RC, Nicoll RA, Mucke L (1999) Plaque-
Res Rev 32:65–74. https​://doi.org/10.1016/j.arr.2016.05.001 independent disruption of neural circuits in Alzheimer’s disease
19. Englund H, Sehlin D, Johansson A-SS, Nilsson LN, Geller- mouse models. Proc Natl Acad Sci USA 96:3228–3233
fors P, Paulie S, Lannfelt L, Pettersson FE (2007) Sensitive 33. Joshi P, Turola E, Ruiz A, Bergami A, Libera DD, Benussi L,
ELISA detection of amyloid-beta protofibrils in biological Giussani P, Magnani G, Comi G, Legname G et al (2014) Micro-
samples. J Neurochem 103:334–345. https​: //doi.org/10.111 glia convert aggregated amyloid-β into neurotoxic forms through
1/j.1471-4159.2007.04759​.x the shedding of microvesicles. Cell Death Differ 21:582–593.
20. Falk A, Koch P, Kesavan J, Takashima Y, Ladewig J, Alexander https​://doi.org/10.1038/cdd.2013.180
M, Wiskow O, Tailor J, Trotter M, Pollard S et al (2012) Capture 34. Kumari S, Mg S, Mayor S (2010) Endocytosis unplugged: mul-
of neuroepithelial-like stem cells from pluripotent stem cells pro- tiple ways to enter the cell. Cell Res 20:256–275. https​://doi.
vides a versatile system for in vitro production of human neurons. org/10.1038/cr.2010.19

13

56 Acta Neuropathologica (2018) 136:41–56

35. Lane RE, Korbie D, Anderson W, Vaidyanathan R, Trau M (2015) intercellular communication in cancer. Onco Targets Ther 7:1327–
Analysis of exosome purification methods using a model liposome 1338. https​://doi.org/10.2147/OTT.S6156​2
system and tunable-resistive pulse sensing. Sci Rep 5:7639. https​ 49. Saman S, Kim W, Raya M, Visnick Y, Miro S, Saman S, Jack-
://doi.org/10.1038/srep0​7639 son B, McKee AC, Alvarez VE, Lee NC et al (2012) Exosome-
36. Logozzi M, De Milito A, Lugini L, Borghi M, Calabro L, Spada associated tau is secreted in tauopathy models and is selectively
M, Perdicchio M, Marino ML, Federici C, Iessi E et al (2009) phosphorylated in cerebrospinal fluid in early Alzheimer dis-
High levels of exosomes expressing CD63 and caveolin-1 in ease. J Biol Chem 287:3842–3849. https​://doi.org/10.1074/jbc.
plasma of melanoma patients. PLoS One 4:e5219. https​://doi. M111.27706​1
org/10.1371/journ​al.pone.00052​19 50. Selkoe DJ (2008) Soluble oligomers of the amyloid beta-pro-
37. Maas SLN, De Vrij J, Broekman MLD (2014) Quantification and tein impair synaptic plasticity and behavior. Behav Brain Res
size-profiling of extracellular vesicles using tunable resistive pulse 192:106–113. https​://doi.org/10.1016/j.bbr.2008.02.016
sensing. Jove-J Vis Exp. https​://doi.org/10.3791/51623​ 51. Stokin GB, Lillo C, Falzone TL, Brusch RG, Rockenstein E,
38. McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Mount SL, Raman R, Davies P, Masliah E, Williams DS et al
Vbeyreuther K, Bush AI, Masters CL (1999) Soluble pool (2005) Axonopathy and transport deficits early in the pathogen-
of Abeta amyloid as a determinant of severity of neurode- esis of alzheimer’s disease. Science 307:1282–1288. https​://doi.
generation in Alzheimer’s disease. Ann Neurol 46:860–866. org/10.1126/scien​ce.11056​81
https ​ : //doi.org/10.1002/1531-8249(19991 ​ 2 )46:6%3C860​ 52. Thal DR, Rub U, Orantes M, Braak H (2002) Phases of a beta-
::AID-ANA8%3E3.0.CO;2-M deposition in the human brain and its relevance for the develop-
39. Narasimhan S, Guo JL, Changolkar L, Stieber A, McBride JD, ment of AD. Neurology 58:1791–1800. https​://doi.org/10.1212/
Silva LV, He Z, Zhang B, Gathagan RJ, Trojanowski JQ et al WNL.58.12.1791
(2017) Pathological tau strains from human brains recapitu- 53. Thal DR, Rub U, Schultz C, Sassin I, Ghebremedhin E, Del
late the diversity of tauopathies in nontransgenic mouse brain. Tredici K, Braak E, Braak H (2000) Sequence of Abeta-protein
J Neurosci 37:11406–11423. https​://doi.org/10.1523/JNEUR​ deposition in the human medial temporal lobe. J Neuropathol Exp
OSCI.1230-17.2017 Neurol 59:733–748. https​://doi.org/10.1093/jnen/59.8.733
40. Nath S, Agholme L, Kurudenkandy FR, Granseth B, Marcus- 54. Thery C, Amigorena S, Raposo G, Clayton A (2006) Isolation
son J, Hallbeck M (2012) Spreading of neurodegenerative and characterization of exosomes from cell culture supernatants
pathology via neuron-to-neuron transmission of beta-amyloid. and biological fluids. Curr Protoc Cell Biol 3:22. https​://doi.
J Neurosci 32:8767–8777. https ​ : //doi.org/10.1523/JNEUR​ org/10.1002/04711​43030​.cb032​2s30
OSCI.0615-12.2012 55. Thery C, Zitvogel L, Amigorena S (2002) Exosomes: composi-
41. Ngolab J, Trinh I, Rockenstein E, Mante M, Florio J, Trejo M, tion, biogenesis and function. Nat Rev Immunol 2:569–579. https​
Masliah D, Adame A, Masliah E, Rissman RA (2017) Brain- ://doi.org/10.1038/nri85​5
derived exosomes from dementia with Lewy bodies propagate 56. Tucker S, Moller C, Tegerstedt K, Lord A, Laudon H, Sjodahl
α-synuclein pathology. Acta Neuropathol Commun 5:46. https​:// J, Soderberg L, Spens E, Sahlin C, Waara ER et al (2015) The
doi.org/10.1186/s4047​8-017-0445-5 murine version of BAN2401 (mAb158) selectively reduces amy-
42. Perez-Gonzalez R, Gauthier SA, Kumar A, Levy E (2012) The loid-beta protofibrils in brain and cerebrospinal fluid of tg-ArcSwe
exosome secretory pathway transports amyloid precursor pro- mice. J Alzheimers Dis 43:575–588. https:​ //doi.org/10.3233/JAD-
tein carboxyl-terminal fragments from the cell into the brain 14074​1
extracellular space. J Biol Chem 287:43108–43115. https​://doi. 57. Tucker S, Möller C, Tegerstedt K, Lord A, Laudon H, Sjödahl
org/10.1074/jbc.M112.40446​7 J, Söderberg L, Spens E, Sahlin C, Waara ER et al (2015) The
43. Pigino G, Morfini G, Atagi Y, Deshpande A, Yu C, Jungbauer L, murine version of BAN2401 (mAb158) selectively reduces
LaDu M, Busciglio J, Brady S (2009) Disruption of fast axonal amyloid-β protofibrils in brain and cerebrospinal fluid of tg-Arc-
transport is a pathogenic mechanism for intraneuronal amy- Swe mice. JAD 43:575–588. https​://doi.org/10.3233/JAD-14074​
loid beta. Proc Natl Acad Sci USA 106:5907–5912. https​://doi. 1
org/10.1073/pnas.09012​29106​ 58. Urbanelli L, Magini A, Buratta S, Brozzi A, Sagini K, Polchi A,
44. Polanco J, Li C, Durisic N, Sullivan R, Götz J (2018) Exosomes Tancini B, Emiliani C (2013) Signaling pathways in exosomes
taken up by neurons hijack the endosomal pathway to spread to biogenesis, secretion and fate. Genes (Basel) 4:152–170. https​://
interconnected neurons. Acta Neuropathol Commun 6:10. https​ doi.org/10.3390/genes​40201​52
://doi.org/10.1186/s4047​8-018-0514-4 59. Vella LJ, Hill AF, Cheng L (2016) Focus on extracellular vesi-
45. Raj A, Kuceyeski A, Weiner M (2012) A network diffusion model cles: exosomes and their role in protein trafficking and biomarker
of disease progression in dementia. Neuron 73:1204–1215. https​ potential in alzheimer’s and parkinson’s disease. Int J Mol Sci
://doi.org/10.1016/j.neuro​n.2011.12.040 17:173. https​://doi.org/10.3390/ijms1​70201​73
46. Rajendran L, Honsho M, Zahn TR, Keller P, Geiger KD, Verkade 60. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe
P, Simons K (2006) Alzheimer’s disease beta-amyloid peptides MS, Rowan MJ, Selkoe DJ (2002) Naturally secreted oligom-
are released in association with exosomes. Proc Natl Acad Sci ers of amyloid beta protein potently inhibit hippocampal long-
USA 103:11172–11177. https:​ //doi.org/10.1073/pnas.060383​ 8103​ term potentiation in  vivo. Nature 416:535–539. https​: //doi.
47. Saheki Y, De Camilli P (2012) Synaptic vesicle endocytosis. Cold org/10.1038/41653​5a
Spring Harb Perspect Biol 4:a005645. https​://doi.org/10.1101/ 61. Walsh DM, Selkoe DJ (2004) Deciphering the molecular basis of
cshpe​rspec​t.a0056​45 memory failure in Alzheimer’s disease. Neuron 44:181–193. https​
48. Salido-Guadarrama I, Romero-Cordoba S, Peralta-Zaragoza ://doi.org/10.1016/j.neuro​n.2004.09.010
O, Hidalgo-Miranda A, Rodriguez-Dorantes M (2014) Micro-
RNAs transported by exosomes in body fluids as mediators of

13

You might also like